1
|
Chen Z, Lan R, Ran T, Tao L, Zhu Y, Li Y, Zhang C, Mao M, Gao D, Zuo Z. A multimodality score strategy for assessing the risk of immune checkpoint inhibitors related cardiotoxicity. Sci Rep 2024; 14:24821. [PMID: 39438579 PMCID: PMC11496699 DOI: 10.1038/s41598-024-76829-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
This study aimed to find the association between four common clinical biomarkers and subsequent ICICT, developing a risk scoring strategy to assess the ICICT risk. Three terminals for ICICT were : Terminal 1, cancer therapy-related cardiomyopathies; Terminal 2, myocarditis or heart failure; and Terminal 3, myocarditis, heart failure, myocardial infarction, cerebral infarction, atrial fibrillation, or death. The thresholds were : N-terminal-pro-B-type-natriuretic-peptide ≥ 125 pg/mL, cardiac troponin T ≥ 6 ng/L, high-sensitivity C-reactive protein ≥ 3 mg/L, and coronary artery calcium score > 10 U. Each of the four abnormal biomarkers received 1 point. The links between biomarkers, score stage, and ICICT were analyzed. 375 patients with a mean follow-up of 1.91 years were included. All four biomarkers measured before immunotherapy were associated with a higher risk of developing ICICT. These scores were also associated with ICICT risk. The highest risk was the very high stage (score = 4) has 7.29, 8.83, and 7.02 folder higher risk compared to low risk group for Terminal 1-3, respectively. The cumulation of incidences also showed that the higher stages of score had an earlier onset and higher incidence of ICICT. 4 biomarkers and the scoring strategy enables clinicians to assess risk easily.
Collapse
Affiliation(s)
- Zhulu Chen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Street, Yuzhong District, Chongqing, 400016, China
| | - Rui Lan
- Department of Clinical Nutrition, School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| | - Tao Ran
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Street, Yuzhong District, Chongqing, 400016, China
| | - Li Tao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxi Zhu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanwei Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Street, Yuzhong District, Chongqing, 400016, China
| | - Chuan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Street, Yuzhong District, Chongqing, 400016, China
| | - Min Mao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Street, Yuzhong District, Chongqing, 400016, China
| | - Diansa Gao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Street, Yuzhong District, Chongqing, 400016, China
| | - Zhong Zuo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Street, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
2
|
Scalia IG, Gheyath B, Tamarappoo BK, Moudgil R, Otton J, Pereyra M, Narayanasamy H, Larsen C, Herrmann J, Arsanjani R, Ayoub C. Chemotherapy Related Cardiotoxicity Evaluation-A Contemporary Review with a Focus on Cardiac Imaging. J Clin Med 2024; 13:3714. [PMID: 38999280 PMCID: PMC11242267 DOI: 10.3390/jcm13133714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The long-term survivorship of patients diagnosed with cancer has improved due to accelerated detection and rapidly evolving cancer treatment strategies. As such, the evaluation and management of cancer therapy related complications has become increasingly important, including cardiovascular complications. These have been captured under the umbrella term "cardiotoxicity" and include left ventricular dysfunction and heart failure, acute coronary syndromes, valvular abnormalities, pericardial disease, arrhythmia, myocarditis, and vascular complications. These complications add to the burden of cardiovascular disease (CVD) or are risk factors patients with cancer treatment are presenting with. Of note, both pre- and newly developing CVD is of prognostic significance, not only from a cardiovascular perspective but also overall, potentially impacting the level of cancer therapy that is possible. Currently, there are varying recommendations and practices regarding CVD risk assessment and mitigating strategies throughout the cancer continuum. This article provides an overview on this topic, in particular, the role of cardiac imaging in the care of the patient with cancer. Furthermore, it summarizes the current evidence on the spectrum, prevention, and management of chemotherapy-related adverse cardiac effects.
Collapse
Affiliation(s)
- Isabel G. Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Bashaer Gheyath
- Department of Imaging, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Balaji K. Tamarappoo
- Division of Cardiology, Banner University Medical Center, The University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Rohit Moudgil
- Department of Cardiology, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James Otton
- Clinical School, St. Vincent’s Hospital, UNSW, Sydney, NSW 2010, Australia
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Hema Narayanasamy
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Carolyn Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| |
Collapse
|
3
|
Murtagh G, Januzzi JL, Scherrer‐Crosbie M, Neilan TG, Dent S, Ho JE, Appadurai V, McDermott R, Akhter N. Circulating Cardiovascular Biomarkers in Cancer Therapeutics-Related Cardiotoxicity: Review of Critical Challenges, Solutions, and Future Directions. J Am Heart Assoc 2023; 12:e029574. [PMID: 37889193 PMCID: PMC10727390 DOI: 10.1161/jaha.123.029574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023]
Abstract
Cardiotoxicity is a growing concern in the oncology population. Transthoracic echocardiography and multigated acquisition scans have been used for surveillance but are relatively insensitive and resource intensive. Innovative imaging techniques are constrained by cost and availability. More sensitive, cost-effective cardiotoxicity surveillance strategies are needed. Circulating cardiovascular biomarkers could provide a sensitive, low-cost solution. Biomarkers such as troponins, natriuretic peptides (NPs), novel upstream signals of oxidative stress, inflammation, and fibrosis as well as panomic technologies have shown substantial promise, and guidelines recommend baseline measurement of troponins and NPs in all patients receiving potential cardiotoxins. Nonetheless, supporting evidence has been hampered by several limitations. Previous reviews have provided valuable perspectives on biomarkers in cancer populations, but important analytic aspects remain to be examined in depth. This review provides comprehensive assessment of critical challenges and solutions in this field, with focus on analytical issues relating to biomarker measurement and interpretation. Examination of evidence pertaining to common and serious forms of cardiotoxicity reveals that improved study designs incorporating larger, more diverse populations, registry-based approaches, and refinement of current definitions are key. Further efforts to harmonize biomarker methodologies including centralized biobanking and analyses, novel decision limits, and head-to-head comparisons are needed. Multimarker algorithms incorporating machine learning may allow rapid, personalized risk assessment. These improvements will not only augment the predictive value of circulating biomarkers in cardiotoxicity but may elucidate both direct and indirect relationships between cardiovascular disease and cancer, allowing biomarkers a greater role in the development and success of novel anticancer therapies.
Collapse
Affiliation(s)
| | - James L. Januzzi
- Division of Cardiology, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | | | - Tomas G. Neilan
- Division of Cardiology, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Susan Dent
- Duke Cancer Institute, Department of MedicineDuke UniversityDurhamNCUSA
| | - Jennifer E. Ho
- CardioVascular Institute and Division of Cardiology, Department of MedicineBeth Israel Deaconess Medicine CenterBostonMAUSA
| | - Vinesh Appadurai
- Division of Cardiovascular MedicineNorthwestern University Feinberg School of MedicineChicagoILUSA
- School of MedicineThe University of QueenslandSt LuciaQueenslandAustralia
| | - Ray McDermott
- Medical OncologySt. Vincent’s University HospitalDublinIreland
| | - Nausheen Akhter
- Division of Cardiovascular MedicineNorthwestern University Feinberg School of MedicineChicagoILUSA
| |
Collapse
|
4
|
Liu C, Chen H, Guo S, Liu Q, Chen Z, Huang H, Zhao Q, Li L, Cen H, Jiang Z, Luo Q, Chen X, Zhao J, Chen W, Yang PC, Wang L. Anti-breast cancer-induced cardiomyopathy: Mechanisms and future directions. Biomed Pharmacother 2023; 166:115373. [PMID: 37647693 DOI: 10.1016/j.biopha.2023.115373] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023] Open
Abstract
With the progression of tumor treatment, the 5-year survival rate of breast cancer is close to 90%. Cardiovascular toxicity caused by chemotherapy has become a vital factor affecting the survival of patients with breast cancer. Anthracyclines, such as doxorubicin, are still some of the most effective chemotherapeutic agents, but their resulting cardiotoxicity is generally considered to be progressive and irreversible. In addition to anthracyclines, platinum- and alkyl-based antitumor drugs also demonstrate certain cardiotoxic effects. Targeted drugs have always been considered a relatively safe option. However, in recent years, some random clinical trials have observed the occurrence of subclinical cardiotoxicity in targeted antitumor drug users, which may be related to the effects of targeted drugs on the angiotensin converting enzyme, angiotensin receptor and β receptor. The use of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and beta-blockers may prevent clinical cardiotoxicity. This article reviews the toxicity and mechanisms of current clinical anti-breast cancer drugs and proposes strategies for preventing cardiovascular toxicity to provide recommendations for the clinical prevention and treatment of chemotherapy-related cardiomyopathy.
Collapse
Affiliation(s)
- Chunping Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China; Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510080, Guangdong Province, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Huiqi Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Sien Guo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Qiaojing Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Zhijun Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Haiding Huang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Qi Zhao
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, Guangdong Province, China
| | - Longmei Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Huan Cen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Qiyuan Luo
- Health Science Center, Shenzhen University, Shenzhen 518060, Guangdong Province, China
| | - Xiaoling Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Jiaxiong Zhao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Wensheng Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Phillip C Yang
- Cardiovascular Stem Cell (Yang) Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China; Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China.
| |
Collapse
|
5
|
Alexandraki A, Papageorgiou E, Zacharia M, Keramida K, Papakonstantinou A, Cipolla CM, Tsekoura D, Naka K, Mazzocco K, Mauri D, Tsiknakis M, Manikis GC, Marias K, Marcou Y, Kakouri E, Konstantinou I, Daniel M, Galazi M, Kampouroglou E, Ribnikar D, Brown C, Karanasiou G, Antoniades A, Fotiadis D, Filippatos G, Constantinidou A. New Insights in the Era of Clinical Biomarkers as Potential Predictors of Systemic Therapy-Induced Cardiotoxicity in Women with Breast Cancer: A Systematic Review. Cancers (Basel) 2023; 15:3290. [PMID: 37444400 PMCID: PMC10340234 DOI: 10.3390/cancers15133290] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Cardiotoxicity induced by breast cancer therapies is a potentially serious complication associated with the use of various breast cancer therapies. Prediction and better management of cardiotoxicity in patients receiving chemotherapy is of critical importance. However, the management of cancer therapy-related cardiac dysfunction (CTRCD) lacks clinical evidence and is based on limited clinical studies. AIM To provide an overview of existing and potentially novel biomarkers that possess a promising predictive value for the early and late onset of CTRCD in the clinical setting. METHODS A systematic review of published studies searching for promising biomarkers for the prediction of CTRCD in patients with breast cancer was undertaken according to PRISMA guidelines. A search strategy was performed using PubMed, Google Scholar, and Scopus for the period 2013-2023. All subjects were >18 years old, diagnosed with breast cancer, and received breast cancer therapies. RESULTS The most promising biomarkers that can be used for the development of an alternative risk cardiac stratification plan for the prediction and/or early detection of CTRCD in patients with breast cancer were identified. CONCLUSIONS We highlighted the new insights associated with the use of currently available biomarkers as a standard of care for the management of CTRCD and identified potentially novel clinical biomarkers that could be further investigated as promising predictors of CTRCD.
Collapse
Affiliation(s)
- Alexia Alexandraki
- A.G. Leventis Clinical Trials Unit, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (E.P.); (M.Z.)
| | - Elisavet Papageorgiou
- A.G. Leventis Clinical Trials Unit, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (E.P.); (M.Z.)
| | - Marina Zacharia
- A.G. Leventis Clinical Trials Unit, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (E.P.); (M.Z.)
| | - Kalliopi Keramida
- 2nd Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
- Cardiology Department, General Anti-Cancer Oncological Hospital, Agios Savvas, 11522 Athens, Greece
| | - Andri Papakonstantinou
- Department of Oncology-Pathology, Karolinska Institute, 17176 Stockholm, Sweden;
- Department for Breast, Endocrine Tumours and Sarcoma, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Carlo M. Cipolla
- Cardioncology and Second Opinion Division, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| | - Dorothea Tsekoura
- 2nd Department of Surgery, Aretaieio University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Av., 11528 Athens, Greece; (D.T.); (E.K.)
| | - Katerina Naka
- 2nd Cardiology Department, University of Ioannina Medical School, 45110 Ioannina, Greece;
| | - Ketti Mazzocco
- Applied Research Division for Cognitive and Psychological Science, European Institute of Oncology IRCCS, 20139 Milan, Italy;
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Davide Mauri
- Department of Medical Oncology, University of Ioannina, 45110 Ioannina, Greece;
| | - Manolis Tsiknakis
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, 71410 Heraklion, Greece; (M.T.); (K.M.)
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece;
| | - Georgios C. Manikis
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece;
| | - Kostas Marias
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, 71410 Heraklion, Greece; (M.T.); (K.M.)
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece;
| | - Yiola Marcou
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Eleni Kakouri
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Ifigenia Konstantinou
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Maria Daniel
- Department of Radiation Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus;
| | - Myria Galazi
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Effrosyni Kampouroglou
- 2nd Department of Surgery, Aretaieio University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Av., 11528 Athens, Greece; (D.T.); (E.K.)
| | - Domen Ribnikar
- Division of Medical Oncology, Institute of Oncology Ljubljana, Faculty of Medicine, University of Ljubljana, Zaloska Cesta 2, 1000 Ljubljana, Slovenia;
| | - Cameron Brown
- Translational Medicine, Stremble Ventures Ltd., 59 Christaki Kranou, Limassol 4042, Cyprus;
| | - Georgia Karanasiou
- Biomedical Research Institute, Foundation for Research and Technology, Hellas, 45500 Ioannina, Greece;
| | - Athos Antoniades
- Research and Development, Stremble Ventures Ltd., 59 Christaki Kranou, Limassol 4042, Cyprus;
| | - Dimitrios Fotiadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece;
| | - Gerasimos Filippatos
- Cardio-Oncology Clinic, Heart Failure Unit, Department of Cardiology, National and Kapodistrian University of Athens Medical School, Athens University Hospital Attikon, 11527 Athens, Greece;
| | - Anastasia Constantinidou
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
- School of Medicine, University of Cyprus, Panepistimiou 1, Aglantzia, Nicosia 2408, Cyprus
| |
Collapse
|
6
|
Al-Kenany SA, Al-Shawi NN. Protective effect of cafestol against doxorubicin-induced cardiotoxicity in rats by activating the Nrf2 pathway. Front Pharmacol 2023; 14:1206782. [PMID: 37377932 PMCID: PMC10291064 DOI: 10.3389/fphar.2023.1206782] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Doxorubicin (DOX) is an efficient antineoplastic agent with a broad antitumor spectrum; however, doxorubicin-associated cardiotoxic adverse effect through oxidative damage and apoptosis limits its clinical application. Cafestol (Caf) is a naturally occurring diterpene in unfiltered coffee with unique antioxidant, antimutagenic, and anti-inflammatory activities by activating the Nrf2 pathway. The present study aimed to investigate the potential chemoprotective effect of cafestol on DOX-induced cardiotoxicity in rats. Wistar albino rats of both sexes were administered cafestol (5 mg/kg/day) for 14 consecutive days by oral gavage alone or with doxorubicin which was injected as a single dose (15 mg/kg intraperitoneally at day 14) to induce toxicity. The result showed that Caf significantly improved cardiac injury induced by doxorubicin, decreased serum levels of CK-MB, LDH, ALP, and ALT, and improved histopathological changes. In addition, cafestol significantly inhibited DOX-induced cardiac oxidative stress as seen in the reduced level of MDA and increased GSH, SOD, CAT, and Gpx-1 cardiac tissue levels; cafestol significantly enhanced Nrf2 gene and protein expression and promoted the expression of downstream antioxidant genes HO-1 and NQO-1 and downregulated Keap1 and NF-κB genes' expression; in addition, Caf significantly reduced inflammatory mediators, TNF-α, and IL-1β levels and inhibited cardiac apoptosis by modulating Bax and Casp 3 tissue levels and reduced TUNEL-positive cardiomyocytes. In conclusion, the present study confirmed that cafestol improved the cardiotoxic effects induced by doxorubicin through the regulation of apoptosis and oxidative stress response through the Nrf2 pathway; this study suggests that cafestol may serve as a potential adjuvant in chemotherapy to alleviate DOX-induced toxicities.
Collapse
|
7
|
Hill L, Delgado B, Lambrinou E, Mannion T, Harbinson M, McCune C. Risk and Management of Patients with Cancer and Heart Disease. Cardiol Ther 2023; 12:227-241. [PMID: 36757637 PMCID: PMC10209380 DOI: 10.1007/s40119-023-00305-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/13/2023] [Indexed: 02/10/2023] Open
Abstract
Cancer and cardiovascular disease are two of the leading causes of global mortality and morbidity. Medical research has generated powerful lifesaving treatments for patients with cancer; however, such treatments may sometimes be at the expense of the patient's myocardium, leading to heart failure. Anti-cancer drugs, including anthracyclines, can result in deleterious cardiac effects, significantly impacting patients' functional capacity, mental well-being, and quality of life. Recognizing this, recent international guidelines and expert papers published recommendations on risk stratification and care delivery, including that of cardio-oncology services. This review will summarize key evidence with a focus on anthracycline therapy, providing clinical guidance for the non-oncology professional caring for a patient with cancer and heart failure.
Collapse
Affiliation(s)
- Loreena Hill
- School of Nursing and Midwifery, Queen's University, 97 Lisburn Road, Belfast, BT9 7BL, UK.
- College of Nursing and Midwifery, Mohammed Bin Rashid University, Dubai, United Arab Emirates.
| | - Bruno Delgado
- Cardiology Department, University Hospital Centre of Oporto, Stº António Hospital, Oporto, Portugal
- Institute of Health Sciences, Portuguese Catholic University, Oporto, Portugal
| | | | - Tara Mannion
- Beaumont Hospital, Dublin, Ireland
- School of Nursing, Midwifery and Health Systems, University College Dublin, Dublin, Ireland
| | - Mark Harbinson
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Claire McCune
- School of Medicine Dentistry and Biomedical Sciences, Queen's University, Belfast, UK
- Belfast Health and Social Care Trust, Belfast, UK
| |
Collapse
|
8
|
Du J, Sudlow LC, Shahverdi K, Zhou H, Michie M, Schindler TH, Mitchell JD, Mollah S, Berezin MY. Oxaliplatin-induced cardiotoxicity in mice is connected to the changes in energy metabolism in the heart tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542198. [PMID: 37292714 PMCID: PMC10245950 DOI: 10.1101/2023.05.24.542198] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oxaliplatin is a platinum-based alkylating chemotherapeutic agent used for cancer treatment. At high cumulative dosage, the negative effect of oxaliplatin on the heart becomes evident and is linked to a growing number of clinical reports. The aim of this study was to determine how chronic oxaliplatin treatment causes the changes in energy-related metabolic activity in the heart that leads to cardiotoxicity and heart damage in mice. C57BL/6 male mice were treated with a human equivalent dosage of intraperitoneal oxaliplatin (0 and 10 mg/kg) once a week for eight weeks. During the treatment, mice were followed for physiological parameters, ECG, histology and RNA sequencing of the heart. We identified that oxaliplatin induces strong changes in the heart and affects the heart's energy-related metabolic profile. Histological post-mortem evaluation identified focal myocardial necrosis infiltrated with a small number of associated neutrophils. Accumulated doses of oxaliplatin led to significant changes in gene expression related to energy related metabolic pathways including fatty acid (FA) oxidation, amino acid metabolism, glycolysis, electron transport chain, and NAD synthesis pathway. At high accumulative doses of oxaliplatin, the heart shifts its metabolism from FAs to glycolysis and increases lactate production. It also leads to strong overexpression of genes in NAD synthesis pathways such as Nmrk2. Changes in gene expression associated with energy metabolic pathways can be used to develop diagnostic methods to detect oxaliplatin-induced cardiotoxicity early on as well as therapy to compensate for the energy deficit in the heart to prevent heart damage.
Collapse
Affiliation(s)
- Junwei Du
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
- Institute of Materials Science & Engineering Washington University, St. Louis, MO 63130, USA
| | - Leland C Sudlow
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Kiana Shahverdi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Haiying Zhou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Megan Michie
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Thomas H Schindler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Joshua D Mitchell
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamim Mollah
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Mikhail Y Berezin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
- Institute of Materials Science & Engineering Washington University, St. Louis, MO 63130, USA
| |
Collapse
|
9
|
Dean M, Kim MJ, Dimauro S, Tannenbaum S, Graham G, Liang BT, Kim AS. Cardiac and noncardiac biomarkers in patients undergoing anthracycline chemotherapy - a prospective analysis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:23. [PMID: 37106424 PMCID: PMC10133897 DOI: 10.1186/s40959-023-00174-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND Biomarkers represent a potential tool to identify individuals at risk for anthracycline-induced cardiotoxicity (AICT) prior to symptom onset or left ventricular dysfunction. METHODS This study examined the levels of cardiac and noncardiac biomarkers before, after the last dose of, and 3-6 months after completion of doxorubicin chemotherapy. Cardiac biomarkers included 5th generation high-sensitivity cardiac troponin T (cTnT), N-terminal pro-brain natriuretic peptide, growth/differentiation factor-15 (GDF-15), and soluble suppression of tumorigenesis-2 (sST2). Noncardiac biomarkers included activated caspase-1 (CASP-1), activated caspase-3, C-reactive protein, tumor necrosis factor-α, myeloperoxidase (MPO), galectin-3, and 8-hydroxy-2'-deoxyguanosine. Echocardiographic data (LVEF and LVGLS) were obtained at pre- and post-chemotherapy. Subanalysis examined interval changes in biomarkers among high (cumulative doxorubicin dose ≥ 250 mg/m2) and low exposure groups. RESULTS The cardiac biomarkers cTnT, GDF-15, and sST2 and the noncardiac biomarkers CASP-1 and MPO demonstrated significant changes over time. cTnT and GDF-15 levels increased after anthracycline exposure, while CASP-1 and MPO decreased significantly. Subanalysis by cumulative dose did not demonstrate a larger increase in any biomarker in the high-dose group. CONCLUSIONS The results identify biomarkers with significant interval changes in response to anthracycline therapy. Further research is needed to understand the clinical utility of these novel biomarkers.
Collapse
Affiliation(s)
- Matthew Dean
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
- Virginia Commonwealth University Health System Internal Medicine Residency, 1101 E. Marshall St, Richmond, VA 23298 USA
| | - Min Jung Kim
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
- Pat and Jim Calhoun Cardiology Center, UConn Health, 300 UConn Health Boulevard, Farmington, CT USA
| | - Sharon Dimauro
- Pat and Jim Calhoun Cardiology Center, UConn Health, 300 UConn Health Boulevard, Farmington, CT USA
| | - Susan Tannenbaum
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
- Carole & Ray Neag Comprehensive Cancer Center, UConn Health, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Garth Graham
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Bruce T. Liang
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
- Pat and Jim Calhoun Cardiology Center, UConn Health, 300 UConn Health Boulevard, Farmington, CT USA
| | - Agnes S. Kim
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
- Pat and Jim Calhoun Cardiology Center, UConn Health, 300 UConn Health Boulevard, Farmington, CT USA
| |
Collapse
|
10
|
Oikawa M, Ishida T, Takeishi Y. Cancer therapeutics-related cardiovascular dysfunction: Basic mechanisms and clinical manifestation. J Cardiol 2023; 81:253-259. [PMID: 35589463 DOI: 10.1016/j.jjcc.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/01/2023]
Abstract
Although recent advances in cancer treatment improve cancer prognosis, cancer therapeutics-related cardiovascular dysfunction (CTRCD) significantly contributes to the global burden of cardiovascular disease. CTRCD causes two crucial issues: first, premature treatment interruption or discontinuation of chemotherapy; second, the development of congestive heart failure during and after cancer treatment. Thus, early detection and prompt treatment of CTRCD may improve the prognosis in cancer patients. This review covers representative anticancer drugs, including anthracyclines, human epidermal growth factor 2 inhibitors, tyrosine kinase inhibitors, proteasome inhibitors, and immune checkpoint inhibitors. We focus on the molecular mechanisms of CTRCD and various approaches to diagnosis, prevention, monitoring, and treatment.
Collapse
Affiliation(s)
- Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University, 1-Hikarigaoka, Fukushima, Fukushima prefecture 960-1295, Japan.
| | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University, 1-Hikarigaoka, Fukushima, Fukushima prefecture 960-1295, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, 1-Hikarigaoka, Fukushima, Fukushima prefecture 960-1295, Japan
| |
Collapse
|
11
|
Hwang S, Kim SH, Yoo KH, Chung MH, Lee JW, Son KH. Exogenous 8-hydroxydeoxyguanosine attenuates doxorubicin-induced cardiotoxicity by decreasing pyroptosis in H9c2 cardiomyocytes. BMC Mol Cell Biol 2022; 23:55. [DOI: 10.1186/s12860-022-00454-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
AbstractDoxorubicin (DOX), which is widely used in cancer treatment, can induce cardiomyopathy. One of the main mechanisms whereby DOX induces cardiotoxicity involves pyroptosis through the NLR family pyrin domain containing 3 (NLRP3) inflammasome and gasdermin D (GSDMD). Increased NAPDH oxidase (NOX) and oxidative stress trigger pyroptosis. Exogenous 8-hydroxydeoxyguanosine (8-OHdG) decreases reactive oxygen species (ROS) production by inactivating NOX. Here, we examined whether 8-OHdG treatment can attenuate DOX-induced pyroptosis in H9c2 cardiomyocytes. Exposure to DOX increased the peroxidative glutathione redox status and NOX1/2/4, toll-like receptor (TLR)2/4, and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) expression, while an additional 8-OHdG treatment attenuated these effects. Furthermore, DOX induced higher expression of NLRP3 inflammasome components, including NLRP3, apoptosis-associated speck-like protein containing a c-terminal caspase recruitment domain (ASC), and pro-caspase-1. Moreover, it increased caspase-1 activity, a marker of pyroptosis, and interleukin (IL)-1β expression. All these effects were attenuated by 8-OHdG treatment. In addition, the expression of the cardiotoxicity markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was increased by DOX, whereas the increase of ANP and BNP induced by DOX treatment was reversed by 8-OHdG. In conclusion, exogenous 8-OHdG attenuated DOX-induced pyroptosis by decreasing the expression of NOX1/2/3, TLR2/4, and NF-κB. Thus, 8-OHdG may attenuate DOX-induced cardiotoxicity through the inhibition of pyroptosis.
Collapse
|
12
|
Bennati E, Girolami F, Spaziani G, Calabri GB, Favre C, Parrini I, Lucà F, Tamburini A, Favilli S. Cardio-Oncology in Childhood: State of the Art. Curr Oncol Rep 2022; 24:1765-1777. [PMID: 36181610 DOI: 10.1007/s11912-022-01329-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Cardio-oncology is an increasingly important field of cardiology that focuses on the detection, monitoring, and treatment of cardiovascular disease (CVD) occurring during and after oncological treatments. The survival rate for childhood cancer patients has dramatically increased thanks to new treatment protocols and cardiovascular (CV) sequelae represent the third most frequent cause of mortality in surviving patients. This study aims to provide a complete and updated review of all the main aspects of cardio-oncology in childhood and to highlight the critical issues. RECENT FINDINGS The problem of CV complications in childhood cancer survivors raises the need to make an early diagnosis of cardiotoxicity by the new imaging and laboratory techniques in order to intervene promptly and to implement pharmacological strategies and lifestyle changes to reduce or even to prevent cardiac injury. Furthermore, a stratification of CV risk, also including new predisposing factors such as the presence of some genetic mutations, is of paramount importance before undertaking oncological treatments. Besides, a systematic and personalized planning of long-term follow-up is fundamental to ensure a transition from pediatric to adult hospital and to avoid missed or late diagnosis of cardiomyopathy. We reviewed the main risk factors for cardiotoxicity in children, both traditional and emerging ones: the mechanisms of toxicity of both old and new antineoplastic therapies, the techniques for detecting cardiac damage, and the current evidence regarding pharmacological cardioprotection. At the end, we focused our attention on the existing guidelines and strategies about the long-term follow-up of childhood cancer survivors.
Collapse
Affiliation(s)
- Elena Bennati
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy.
| | - Francesca Girolami
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Gaia Spaziani
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | | | - Claudio Favre
- Department of Pediatric Hematology-Oncology, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Iris Parrini
- Cardiology Unit, Mauriziano Umberto I Hospital, Corso Turati 62, Turin, Italy
| | - Fabiana Lucà
- Department of Cardiology, Grande Ospedale Metropolitano, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Angela Tamburini
- Department of Pediatric Hematology-Oncology, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Silvia Favilli
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| |
Collapse
|
13
|
Porter C, Azam TU, Mohananey D, Kumar R, Chu J, Lenihan D, Dent S, Ganatra S, Beasley GS, Okwuosa T. Permissive Cardiotoxicity. JACC CardioOncol 2022; 4:302-312. [PMID: 36213359 PMCID: PMC9537074 DOI: 10.1016/j.jaccao.2022.07.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
The field of cardio-oncology was born from the necessity for recognition and management of cardiovascular diseases among patients with cancer. This need for this specialty continues to grow as patients with cancer live longer as a result of lifesaving targeted and immunologic cancer therapies beyond the usual chemotherapy and/or radiation therapy. Often, potentially cardiotoxic anticancer treatment is necessary in patients with baseline cardiovascular disease. Moreover, patients may need to continue therapy in the setting of incident cancer therapy–associated cardiotoxicity. Herein, we present and discuss the concept of permissive cardiotoxicity as a novel term that represents an essential concept in the field of cardio-oncology and among practicing cardio-oncology specialists. It emphasizes a proactive rather than reactive approach to continuation of lifesaving cancer therapies in order to achieve the best oncologic outcome while mitigating associated and potentially off-target cardiotoxicities. Permissive cardiotoxicity is a terminology that represents a vital concept in cardio-oncology It emphasizes continued cancer therapy if appropriate, while mitigating cardiotoxicities. Its application is guided by understanding the cancer treatment, alternatives, and prognosis.
Collapse
Affiliation(s)
- Charles Porter
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Address for correspondence: Dr Charles Porter, University of Kansas Medical Center, 4000 Cambridge, MS 1023, Kansas City, Kansas 66160, USA. @charlesporter99
| | - Tariq U. Azam
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Divyanshu Mohananey
- Division of Cardiology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Rohit Kumar
- Division of Medical Oncology, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jian Chu
- Department of Medicine, Rush University, Chicago, Illinois, USA
| | - Daniel Lenihan
- International Cardio-Oncology Society, Tampa, Florida, USA
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Gary S. Beasley
- Division of Cardiology, Department of Pediatrics, University of Iowa Stead Family Children’s Hospital, Iowa City, Iowa, USA
| | - Tochukwu Okwuosa
- Division of Cardiology, Department of Internal Medicine, Rush University, Chicago, Illinois, USA
| |
Collapse
|
14
|
Hamid A, Anker MS, Ruckdeschel JC, Khan MS, Tharwani A, Oshunbade AA, Kipchumba RK, Thigpen SC, Anker SD, Fonarow GC, Hall ME, Butler J. Cardiovascular Safety Reporting in Contemporary Breast Cancer Clinical Trials. J Am Heart Assoc 2022; 11:e025206. [PMID: 35876414 PMCID: PMC9375478 DOI: 10.1161/jaha.121.025206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Several cancer therapies have been associated with cardiovascular harm in early‐phase clinical trials. However, some cardiovascular harms do not manifest until later‐phase trials. To limit interdisease variability, we focused on breast cancer. Thus, we assessed the reporting of cardiovascular safety monitoring and outcomes in phase 2 and 3 contemporary breast cancer clinical trials. Methods and Results We searched Embase and Medline records for phase 2 and 3 breast cancer pharmacotherapy trials. We examined exclusion criterion as a result of cardiovascular conditions, adverse cardiovascular event reporting, and cardiovascular safety assessment through cardiovascular imaging, ECG, troponin, or natriuretic peptides. Fisher's exact test was utilized to compare reporting. Fifty clinical trials were included in our study. Patients were excluded because of cardiovascular conditions in 42 (84%) trials. Heart failure was a frequent exclusion criterion (n=31; 62% trials). Adverse cardiovascular events were reported in 43 (86%) trials. Cardiovascular safety assessments were not reported in 23 (46%) trials, whereas natriuretic peptide and troponin assessments were not reported in any trial. Cardiovascular safety assessments were more frequently reported in industry‐funded trials (69.2% versus 0.0%; P<0.001), and in trials administering targeted/immunotherapy agents compared with only hormonal/conventional chemotherapy (78.6% versus 22.7%, P<0.001). Conclusions Our findings demonstrate significant under‐representation of patients with cardiovascular conditions or prevalent cardiovascular disease in contemporary later‐phase breast cancer trials. Additionally, cardiovascular safety is not routinely monitored in these trials. Therefore, contemporary breast cancer clinical trials may possibly underestimate the cardiovascular risks of cancer pharmacotherapy agents for use in clinical practice.
Collapse
Affiliation(s)
- Arsalan Hamid
- Department of Medicine University of Mississippi Medical Center Jackson MS
| | - Markus S Anker
- Department of Cardiology (CBF), Charité University Medicine Berlin Berlin Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT) Berlin Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin Berlin Germany
| | - John C Ruckdeschel
- Division of Hematology/Oncology, Department of Medicine, Cancer Center and Research Institute University of Mississippi Medical Center Jackson MS
| | | | - Arsal Tharwani
- Department of Medicine Cleveland Clinic Foundation Cleveland OH
| | - Adebamike A Oshunbade
- Division of Cardiology, Department of Medicine University of Mississippi Medical Center Jackson MS
| | - Rodney K Kipchumba
- Department of Medicine University of Mississippi Medical Center Jackson MS
| | - Samuel C Thigpen
- Department of Medicine University of Mississippi Medical Center Jackson MS
| | - Stefan D Anker
- Berlin Institute of Health Center for Regenerative Therapies (BCRT) Berlin Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin Berlin Germany.,Department of Cardiology (CVK) Charité Universitätsmedizin Berlin Berlin Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT) Charité Universitätsmedizin Berlin Germany.,German Centre for Cardiovascular Research (DZHK) partner site Berlin Charité Universitätsmedizin Berlin Germany
| | - Gregg C Fonarow
- Division of Cardiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Michael E Hall
- Division of Cardiology, Department of Medicine University of Mississippi Medical Center Jackson MS
| | - Javed Butler
- Department of Medicine University of Mississippi Medical Center Jackson MS
| |
Collapse
|
15
|
Tonry C, Russel-Hallinan A, McCune C, Collier P, Harbinson M, Dixon L, Watson CJ. Circulating biomarkers for management of cancer therapeutics related cardiac dysfunction. Cardiovasc Res 2022; 119:710-728. [PMID: 35640873 PMCID: PMC10153425 DOI: 10.1093/cvr/cvac087] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer therapeutics related cardiac dysfunction (CTRCD) has emerged as a major cause of morbidity and mortality in cancer survivors. Effective clinical management of CTRCD is impeded by a lack of sensitive diagnostic and prognostic strategies. Circulating molecular markers could potentially address this need as they are often indicative of cardiac stress before cardiac damage can be detected clinically. A growing understanding of the underlying physiological mechanisms for CTRCD has inspired research efforts to identify novel pathophysiologically-relevant biomarkers that may also guide development of cardio-protective therapeutic approaches. The purpose of this review is to evaluate current circulating biomarkers of cardiac stress and their potential role in diagnosis and management of CTRCD. We also discuss some emerging avenues for CTRCD-focused biomarker investigations.
Collapse
Affiliation(s)
- Claire Tonry
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Adam Russel-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Claire McCune
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | | | | | | | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
16
|
Saha S, Singh PK, Roy P, Kakar SS. Cardiac Cachexia: Unaddressed Aspect in Cancer Patients. Cells 2022; 11:cells11060990. [PMID: 35326441 PMCID: PMC8947289 DOI: 10.3390/cells11060990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/14/2022] Open
Abstract
Tumor-derived cachectic factors such as proinflammatory cytokines and neuromodulators not only affect skeletal muscle but also affect other organs, including the heart, in the form of cardiac muscle atrophy, fibrosis, and eventual cardiac dysfunction, resulting in poor quality of life and reduced survival. This article reviews the holistic approaches of existing diagnostic, pathophysiological, and multimodal therapeutic interventions targeting the molecular mechanisms that are responsible for cancer-induced cardiac cachexia. The major drivers of cardiac muscle wasting in cancer patients are autophagy activation by the cytokine-NFkB, TGF β-SMAD3, and angiotensin II-SOCE-STIM-Ca2+ pathways. A lack of diagnostic markers and standard treatment protocols hinder the early diagnosis of cardiac dysfunction and the initiation of preventive measures. However, some novel therapeutic strategies, including the use of Withaferin A, have shown promising results in experimental models, but Withaferin A’s effectiveness in human remains to be verified. The combined efforts of cardiologists and oncologists would help to identify cost effective and feasible solutions to restore cardiac function and to increase the survival potential of cancer patients.
Collapse
Affiliation(s)
- Sarama Saha
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh 249203, India; (S.S.); (P.K.S.)
| | - Praveen Kumar Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh 249203, India; (S.S.); (P.K.S.)
| | - Partha Roy
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India;
| | - Sham S. Kakar
- Department of Physiology and Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA
- Correspondence: ; Tel.: +1-(502)-852-0812
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Successful treatment of cancer can be hampered by the attendant risk of cardiotoxicity, manifesting as cardiomyopathy, left ventricle systolic dysfunction and, in some cases, heart failure. This risk can be mitigated if the injury to the heart is detected before the onset to irreversible cardiac impairment. The gold standard for cardiac imaging in cardio-oncology is echocardiography. Despite improvements in the application of this modality, it is not typically sensitive to sub-clinical or early-stage dysfunction. We identify in this review some emerging tracers for detecting incipient cardiotoxicity by positron emission tomography (PET). RECENT FINDINGS Vectors labeled with positron-emitting radionuclides (e.g., carbon-11, fluorine-18, gallium-68) are now available to study cardiac function, metabolism, and tissue repair in preclinical models. Many of these probes are highly sensitive to early damage, thereby potentially addressing the limitations of current imaging approaches, and show promise in preliminary clinical evaluations. The overlapping pathophysiology between cardiotoxicity and heart failure significantly expands the number of imaging tools available to cardio-oncology. This is highlighted by the emergence of radiolabeled probes targeting fibroblast activation protein (FAP) for sensitive detection of dysregulated healing process that underpins adverse cardiac remodeling. The growth of PET scanner technology also creates an opportunity for a renaissance in metabolic imaging in cardio-oncology research.
Collapse
Affiliation(s)
- James M. Kelly
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
| | - John W. Babich
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021 USA
| |
Collapse
|
18
|
Abstract
The development of cardiovascular toxicity attributable to anticancer drugs is a pivotal event that is associated with cardiovascular morbidity as well as with worse cancer-specific and overall outcomes. Although broad consensus exists regarding the importance of cardiovascular safety assessment in cancer drug development, real-world data suggest that cardiovascular events are significantly underestimated in oncology trials. This drug safety discrepancy has profound implications on drug development decisions, risk-benefit evaluation, formulation of surveillance and prevention protocols, and survivorship. In this article, we review the contemporary cardiovascular safety evaluation of new pharmaceuticals in hematology and oncology, spanning from in vitro pharmacodynamic testing to randomized clinical trials. We argue that cardiovascular safety assessment of anticancer drugs should be reformed and propose practical strategies, including development and validation of preclinical assays, expansion of oncology trial eligibility, incorporation of cardiovascular end points in early-phase studies, and design of longitudinal multi-institutional cardiotoxicity registries.
Collapse
Affiliation(s)
- Ohad Oren
- Division of CardiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Tomas G. Neilan
- Cardio‐Oncology ProgramDivision of CardiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Michael G. Fradley
- Cardio‐Oncology Center of ExcellenceDivision of CardiologyDepartment of MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPA
| | - Deepak L. Bhatt
- Brigham and Women’s Hospital Heart & Vascular Center and Harvard Medical SchoolBostonMA
| |
Collapse
|
19
|
Zhang C, Chen Z, Mo C, Gao D, Zhu Y, Qin S, Zuo Z. Real-world cardiovascular toxicity of immune checkpoint inhibitors in cancer patients: a retrospective controlled cohort study. Am J Cancer Res 2021; 11:6074-6085. [PMID: 35018243 PMCID: PMC8727793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/21/2021] [Indexed: 06/14/2023] Open
Abstract
Over the past decade, immune checkpoint inhibitors (ICI) have dramatically improved the prognosis of many cancer patients, but many immune-related adverse cardiovascular events (ACEs) have been observed. We aimed to investigate the occurrence of ACEs in the real world after receiving ICI and provide clinical reference for how to evaluate it. The study retrospectively included 204 patients who received ICI from October 2019 to November 2020 and 205 patients who only received traditional chemotherapy. The mean duration of follow-up for ICI group was 4.88 months, and the control group was 4.79 months. Patients in the control group did not develop myocarditis, only 2 cases of new-onset pericardial effusion occurred. In contrast, among ICI group, there were 3 cases of ICI-associated myocarditis, accounting for 1.47% (3/204), 6 cases of pericardial effusion. The incidence of new-onset ECG abnormalities in the ICI group was significantly higher than that of the control group (38/180 VS 16/178, HR 2.71, 95% CI: 1.449-5.067, P=0.001). In the ICI group, after receiving ICI treatment, cardiac biomarkers including average cardiac troponin T and N terminal pro B type natriuretic peptide increased significantly, peak in about 1 month, and then gradually decreasing. After the third or fourth month, the cardiac biomarkers gradually increased again. In conclusion, ICI may lead to various ACEs, and its incidence is higher than that of patients who only receive traditional chemotherapy. The changing trend of cardiac biomarkers reflects that ICI may cause acute and chronic myocardial damage. Regularly performing ECG, echocardiogram and cardiac biomarker examinations are helpful for early detection of ACEs caused by ICI and providing timely treatment.
Collapse
Affiliation(s)
- Chuan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University Youyi Road, Chongqing 400016, China
| | - Zhulu Chen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University Youyi Road, Chongqing 400016, China
| | - Chunhua Mo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University Youyi Road, Chongqing 400016, China
| | - Diansha Gao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University Youyi Road, Chongqing 400016, China
| | - Yuxi Zhu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University Youyi Road, Chongqing 400016, China
| | - Shu Qin
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University Youyi Road, Chongqing 400016, China
| | - Zhong Zuo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University Youyi Road, Chongqing 400016, China
| |
Collapse
|
20
|
Chen D, Kelly C, Haw TJ, Lombard JM, Nordman IIC, Croft AJ, Ngo DTM, Sverdlov AL. Heart Failure in Breast Cancer Survivors: Focus on Early Detection and Novel Biomarkers. Curr Heart Fail Rep 2021; 18:362-377. [PMID: 34731413 DOI: 10.1007/s11897-021-00535-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2021] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW Breast cancer survival rate has greatly improved in the last two decades due to the emergence of next-generation anti-cancer agents. However, cardiotoxicity remains a significant adverse effect arising from traditional and emerging chemotherapies as well as targeted therapies for breast cancer patients. In this review, we will discuss cardiotoxicities of both traditional and emerging therapies for breast cancer. We will discuss current practices to detect cardiotoxicity of these therapies with the focus on new and emerging biomarkers. We will then focus on 'omics approaches, especially the use of epigenetics to discover novel biomarkers and therapeutics to mitigate cardiotoxicity. RECENT FINDINGS Significant cardiotoxicities of conventional chemotherapies remain and new and unpredictable new forms of cardiac and/or vascular toxicity emerge with the surge in novel and targeted therapies. Yet, there is no clear guidance on detection of cardiotoxicity, except for significant left ventricular systolic dysfunction, and even then, there is no uniform definition of what constitutes cardiotoxicity. The gold standard for detection of cardiotoxicity involves a serial echocardiography in conjunction with blood-based biomarkers to detect early subclinical cardiac dysfunction. However, the ability of these tests to detect early disease remains limited and not all forms of toxicity are detectable with these modalities. There is an unprecedented need to discover novel biomarkers that are sensitive and specific for early detection of subclinical cardiotoxicity. In that space, novel echocardiographic techniques, such as strain, are becoming more common-place and new biomarkers, discovered by epigenetic approaches, seem to become promising alternatives or adjuncts to conventional non-specific cardiac biomarkers.
Collapse
Affiliation(s)
- Dongqing Chen
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia
| | - Conagh Kelly
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia
| | - Tatt Jhong Haw
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia.,Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Janine M Lombard
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Ina I C Nordman
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Amanda J Croft
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Doan T M Ngo
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia. .,School of Biomedical Science and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| | - Aaron L Sverdlov
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia. .,Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia. .,Cardiovascular Department, John Hunter Hospital, Hunter New England Local Health District, NSW, New Lambton Heights, Australia. .,School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
21
|
Speers C, Murthy VL, Walker EM, Glide-Hurst CK, Marsh R, Tang M, Morris EL, Schipper MJ, Weinberg RL, Gits HC, Hayman J, Feng M, Balter J, Moran J, Jagsi R, Pierce LJ. Cardiac Magnetic Resonance Imaging and Blood Biomarkers for Evaluation of Radiation-Induced Cardiotoxicity in Patients With Breast Cancer: Results of a Phase 2 Clinical Trial. Int J Radiat Oncol Biol Phys 2021; 112:417-425. [PMID: 34509552 DOI: 10.1016/j.ijrobp.2021.08.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE Radiation therapy (RT) can increase the risk of cardiac events in patients with breast cancer (BC), but biomarkers predicting risk for developing RT-induced cardiac disease are currently lacking. We report results from a prospective clinical trial evaluating early magnetic resonance imaging (MRI) and serum biomarker changes as predictors of cardiac injury and risk of subsequent cardiac events after RT for left-sided disease. METHODS Women with node-negative and node-positive (N-/+) left-sided BC were enrolled on 2 institutional review board (IRB)-approved protocols at 2 institutions. MRI was conducted pretreatment (within 1 week of starting radiation), at the end of treatment (last day of treatment ±1 week), and 3 months after the last day of treatment (±2 weeks) to quantify left and right ventricular volumes and function, myocardial fibrosis, and edema. Perfusion changes during regadenoson stress perfusion were also assessed on a subset of patients (n = 28). Serum was collected at the same time points. Whole heart and cardiac substructures were contoured using CT and MRI. Models were constructed using baseline cardiac and clinical risk factors. Associations between MRI-measured changes and dose were evaluated. RESULTS Among 51 women enrolled, mean heart dose ranged from 0.80 to 4.7 Gy and mean left ventricular (LV) dose from 1.1 to 8.2 Gy, with mean heart dose 2.0 Gy. T1 time, a marker of fibrosis, and right ventricular (RV) ejection fraction (EF) significantly changed with treatment; these were not dose dependent. T2 (marker of edema) and LV EF did not significantly change. No risk factors were associated with baseline global perfusion. Prior receipt of doxorubicin was marginally associated with decreased myocardial perfusion after RT (P = .059), and mean MHD was not associated with perfusion changes. A significant correlation between baseline IL-6 and mean heart dose (MHD) at the end of RT (ρ 0.44, P = .007) and a strong trend between troponin I and MHD at 3 months post-treatment (ρ 0.33, P = .07) were observed. No other significant correlations were identified. CONCLUSIONS In this prospective study of women with left-sided breast cancer treated with contemporary treatment planning, cardiac radiation doses were very low relative to historical doses reported by Darby et al. Although we observed significant changes in T1 and RV EF shortly after RT, these changes were not correlated with whole heart or substructure doses. Serum biomarker analysis of cardiac injury demonstrates an interesting trend between markers and MHD that warrants further investigation.
Collapse
Affiliation(s)
- Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Venkatesh L Murthy
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan
| | - Eleanor M Walker
- Department of Radiation Oncology, Henry Ford Cancer Institute, Detroit, Michigan
| | - Carri K Glide-Hurst
- Department of Human Oncology, School of Medicine and Public Heath, University of Wisconsin-Madison, Madison, Wisconsin
| | - Robin Marsh
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Ming Tang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Emily L Morris
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Matthew J Schipper
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Richard L Weinberg
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan
| | - Hunter C Gits
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - James Hayman
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Mary Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - James Balter
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Jean Moran
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Reshma Jagsi
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Lori J Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
22
|
Saleh Y, Abdelkarim O, Herzallah K, Abela GS. Anthracycline-induced cardiotoxicity: mechanisms of action, incidence, risk factors, prevention, and treatment. Heart Fail Rev 2021; 26:1159-1173. [PMID: 32410142 DOI: 10.1007/s10741-020-09968-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Anthracycline is a mainstay in treatment of many cancers including lymphoma and breast cancer among many others. However, anthracycline treatment can be cardiotoxic. Although anthracycline-induced cardiotoxicity is dose dependent, it can also occur early at the onset of treatment and even up to several years following completion of treatment. This review article focuses on the understanding of mechanisms of anthracycline-induced cardiotoxicity, the treatments, and recommended follow-up and preventive approaches.
Collapse
Affiliation(s)
- Yehia Saleh
- Department of Internal Medicine, Michigan State University, East Lansing, MI, USA
| | - Ola Abdelkarim
- Department of Internal Medicine, Cardiology, Michigan State University, 788 service road, Room B-208, Clinical Center, East Lansing, MI, USA
| | - Khader Herzallah
- Department of Internal Medicine, Michigan State University, East Lansing, MI, USA
| | - George S Abela
- Department of Internal Medicine, Cardiology, Michigan State University, 788 service road, Room B-208, Clinical Center, East Lansing, MI, USA.
| |
Collapse
|
23
|
Radulescu LM, Radulescu D, Ciuleanu TE, Crisan D, Buzdugan E, Romitan DM, Buzoianu AD. Cardiotoxicity Associated with Chemotherapy Used in Gastrointestinal Tumours. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57080806. [PMID: 34441012 PMCID: PMC8400748 DOI: 10.3390/medicina57080806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/25/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
Cardiotoxicity is a well-recognised side effect of cancer-related therapies with a great impact on outcomes and quality of life in the cancer survivor population. The pathogenesis of chemotherapy-induced cardiotoxicity in patients with gastrointestinal cancers involves various molecular mechanisms, and the combined use of various chemotherapies augments the risk of each drug used alone. In terms of cardiotoxicity diagnosis, novel biomarkers, such as troponins, brain natriuretic peptide (BNP), myeloperoxidases and miRNAs have been recently assessed. Echocardiography is a noninvasive imaging method of choice for the primary assessment of chemotherapy-treated patients to generally evaluate the cardiovascular impact of these drugs. Novel echocardiography techniques, like three-dimensional and stress echocardiography, will improve diagnosis efficacy. Cardiac magnetic resonance (CMR) can evaluate cardiac morphology, function and wall structure. Corroborated data have shown the importance of CMR in the early evaluation of patients with gastrointestinal cancers, treated with anticancer drugs, but further studies are required to improve risk stratification in these patients. In this article, we review some important aspects concerning the cardiotoxicity of antineoplastic drugs used in gastrointestinal cancers. We also discuss the mechanism of cardiotoxicity, the role of biomarkers and the imaging methods used in its detection.
Collapse
Affiliation(s)
- Liliana Maria Radulescu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania; (L.M.R.); (A.D.B.)
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
| | - Dan Radulescu
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
- Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania;
- Correspondence: ; Tel.: +40-741041707
| | - Tudor-Eliade Ciuleanu
- Department of Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania;
| | - Dana Crisan
- Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania;
- Department of Internal Medicine, Cluj Municipal Hospital, 400139 Cluj-Napoca, Romania
| | - Elena Buzdugan
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
- Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania;
| | - Dragos-Mihai Romitan
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania; (L.M.R.); (A.D.B.)
| |
Collapse
|
24
|
de Wall C, Bauersachs J, Berliner D. Cardiooncology-dealing with modern drug treatment, long-term complications, and cancer survivorship. Clin Exp Metastasis 2021; 38:361-371. [PMID: 34117981 PMCID: PMC8318956 DOI: 10.1007/s10585-021-10106-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/28/2021] [Indexed: 01/22/2023]
Abstract
Modern treatment strategies have improved prognosis and survival of patients with malignant diseases. The key components of tumor treatment are conventional chemotherapy, radiotherapy, targeted therapies, and immunotherapy. Cardiovascular side-effects may occur in the early phase of tumor therapy or even decades later. Therefore, knowledge and awareness of acute and long-lasting cardiac side effects of anti-cancer therapies are essential. Cardiotoxicity impairs quality of life and overall survival. The new cardiologic subspecialty 'cardio-oncology' deals with the different cardiovascular problems arising from tumor treatment and the relationship between cancer and heart diseases. Early detection and treatment of cardiotoxicity is of crucial importance. A detailed cardiac assessment of patients prior to administration of cardiotoxic agents, during and after treatment should be performed in all patients. The current review focusses on acute and long-term cardiotoxic side effects of classical cytotoxic and selected modern drug treatments such as immune checkpoint inhibitors and discusses strategies for the diagnosis of treatment-related adverse cardiovascular effects in cancer patients.
Collapse
Affiliation(s)
- Claudia de Wall
- Dept. of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Johann Bauersachs
- Dept. of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Dominik Berliner
- Dept. of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
25
|
Bikiewicz A, Banach M, von Haehling S, Maciejewski M, Bielecka‐Dabrowa A. Adjuvant breast cancer treatments cardiotoxicity and modern methods of detection and prevention of cardiac complications. ESC Heart Fail 2021; 8:2397-2418. [PMID: 33955207 PMCID: PMC8318493 DOI: 10.1002/ehf2.13365] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022] Open
Abstract
The most common cancer diagnosis in female population is breast cancer, which affects every year about 2.0 million women worldwide. In recent years, significant progress has been made in oncological therapy, in systemic treatment, and in radiotherapy of breast cancer. Unfortunately, the improvement in the effectiveness of oncological treatment and prolonging patients' life span is associated with more frequent occurrence of organ complications, which are side effects of this treatment. Current recommendations suggest a periodic monitoring of the cardiovascular system in course of oncological treatment. The monitoring includes the assessment of occurrence of risk factors for cardiovascular diseases in combination with the evaluation of the left ventricular systolic function using echocardiography and electrocardiography as well as with the analysis of the concentration of cardiac biomarkers. The aim of this review was critical assessment of the breast cancer therapy cardiotoxicity and the analysis of methods its detections. The new cardio-specific biomarkers in serum, the development of modern imaging techniques (Global Longitudinal Strain and Three-Dimensional Left Ventricular Ejection Fraction) and genotyping, and especially their combined use, may become a useful tool for identifying patients at risk of developing cardiotoxicity, who require further cardiovascular monitoring or cardioprotective therapy.
Collapse
Affiliation(s)
- Agata Bikiewicz
- Heart Failure Unit, Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)Rzgowska 281/289Lodz93‐338Poland
- Department of Hypertension, Chair of Nephrology and HypertensionMedical University of LodzLodzPoland
| | - Maciej Banach
- Heart Failure Unit, Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)Rzgowska 281/289Lodz93‐338Poland
- Department of Hypertension, Chair of Nephrology and HypertensionMedical University of LodzLodzPoland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology and German Center for Cardiovascular Research (DZHK), partner site GöttingenUniversity Medical Center Göttingen (UMG)GöttingenGermany
| | - Marek Maciejewski
- Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)LodzPoland
| | - Agata Bielecka‐Dabrowa
- Heart Failure Unit, Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)Rzgowska 281/289Lodz93‐338Poland
- Department of Hypertension, Chair of Nephrology and HypertensionMedical University of LodzLodzPoland
| |
Collapse
|
26
|
Kim SR, Heo JI, Park JW, Kang CM, Kim KS. Radiation-induced lipoprotein-associated phospholipase A2 increases lysophosphatidylcholine and induces endothelial cell damage. Toxicology 2021; 458:152841. [PMID: 34216699 DOI: 10.1016/j.tox.2021.152841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
The cardiotoxicity of various anticancer therapies, including radiotherapy, can lead to cardiovascular complications. These complications can range from damaging cardiac tissues within the irradiation field to increasing the long-term risks of developing heart failure, coronary artery disease, and myocardial infarction. We analyzed radiation-induced metabolites capable of mediating critical biological processes, such as inflammation, senescence, and apoptosis. Previously, by applying QTOF-MASS analysis to irradiated human fibroblasts, we identified that metabolite sets of lysophosphatidylcholine (LPC) were increased in these cells. In this study, radiation-induced LPC accumulation in human aortic endothelial cells (HAECs) increased reactive oxygen species (ROS) production and senescence-associated-beta-galactosidase staining, in addition to decreasing their tube-forming ability. Knockdown of lipoprotein-associated phospholipase A2 (Lp-PLA2) with small interfering RNA (siRNA) inhibited the increased LPC production induced by radiation, and reduced the radiation-induced cell damage produced by ROS and oxidized low-density lipoprotein (LDL). Lp-PLA2 depletion abolished the induction of proinflammatory factors, such as interleukin 1β, tumor necrosis factor-alpha, matrix metalloproteinase 2, and matrix metalloproteinase 9, as well as adhesion molecules, such as intercellular adhesion molecule 1 (ICAM-1) and E-selection. Likewise, we showed that Lp-PLA2 expression was upregulated in the vasculature of irradiated rat, resulting in increased LPC production and LDL oxidation. Our data demonstrate that radiation-induced LPC production is a potential risk factor for cardiotoxicity that is mediated by Lp-PLA2 activity, suggesting that LPC and Lp-PLA2 offer potential diagnostic and therapeutic approaches to cardiovascular damage during radiotherapy.
Collapse
Affiliation(s)
- So-Ra Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea; School of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34054, Republic of Korea
| | - Jong-Ik Heo
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Jeong-Woo Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Chang-Mo Kang
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea; School of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34054, Republic of Korea.
| | - Kwang Seok Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea; School of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34054, Republic of Korea.
| |
Collapse
|
27
|
Gong FF, Cascino GJ, Murtagh G, Akhter N. Circulating Biomarkers for Cardiotoxicity Risk Prediction. Curr Treat Options Oncol 2021; 22:46. [PMID: 33866434 DOI: 10.1007/s11864-021-00845-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 01/22/2023]
Abstract
OPINION STATEMENT Improvements in cancer survival have led to the emergence of cardiovascular disease as an important determinant of adverse outcome in survivors. Cancer therapeutics-related cardiac dysfunction is the most well-known form of cardiotoxicity. However, newer cancer therapies bring a broader range of cardiotoxicities. The optimal method to identify patients at risk of these complications is unclear, but circulating biomarkers comprise one possible approach. Troponins and natriuretic peptides have garnered the broadest evidence base for cardiotoxicity risk prediction, but other markers are being investigated. In this review, we explore evidence for circulating biomarkers in cardiotoxicity prediction associated with cancer therapies.
Collapse
Affiliation(s)
- Fei Fei Gong
- Division of Cardiology, Northwestern University Feinberg School of Medicine, 676 N St Clair, Suite 600, Chicago, IL, 60611, USA
| | - Gregory J Cascino
- Division of Cardiology, Northwestern University Feinberg School of Medicine, 676 N St Clair, Suite 600, Chicago, IL, 60611, USA
| | - Gillian Murtagh
- Diagnostics Division, Abbott Laboratories, 100 Abbott Park Road, CP1-3, Abbott Park, North Chicago, IL, 60064-6094, USA
| | - Nausheen Akhter
- Division of Cardiology, Northwestern University Feinberg School of Medicine, 676 N St Clair, Suite 600, Chicago, IL, 60611, USA.
| |
Collapse
|
28
|
Crimmin J, Fulop T, Battisti NML. Biological aspects of aging that influence response to anticancer treatments. Curr Opin Support Palliat Care 2021; 15:29-38. [PMID: 33399393 DOI: 10.1097/spc.0000000000000536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Cancer is a disease of older adults, where fitness and frailty are a continuum. This aspect poses unique challenges to the management of cancer in this population. In this article, we review the biological aspects influencing the efficacy and safety of systemic anticancer treatments. RECENT FINDINGS The organ function decline associated with the ageing process affects multiple systems, including liver, kidney, bone marrow, heart, muscles and central nervous system. These can have a significant impact on the pharmacokinetics and pharmacodynamics of systemic anticancer agents. Comorbidities also represent a key aspect to consider in decision-making. Renal disease, liver conditions and cardiovascular risk factors are prevalent in this age group and may impact the risk of adverse outcomes in this setting. SUMMARY The systematic integration of geriatrics principles in the routine management of older adults with cancer is a unique opportunity to address the complexity of this population and is standard of care based on a wide range of benefits. This approach should be multidisciplinary and involve careful discussion with hospital pharmacists.
Collapse
Affiliation(s)
- Jane Crimmin
- Pharmacy, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | - Tamas Fulop
- Department of Medicine, Division of Geriatrics, Research Center on Aging, University of Sherbrooke, Faculty of Medicine and Health Sciences, Québec, Quebec, Canada
| | - Nicolò Matteo Luca Battisti
- Department of Medicine - Breast Unit, The Royal Marsden NHS Foundation Trust, Breast Cancer Research Division, The Institute of Cancer Research, Sutton, Surrey, UK
| |
Collapse
|
29
|
Di Lisi D, Manno G, Novo G. Subclinical Cardiotoxicity: The Emerging Role of Myocardial Work and Other Imaging Techniques. Curr Probl Cardiol 2021; 46:100818. [PMID: 33756178 DOI: 10.1016/j.cpcardiol.2021.100818] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 02/01/2021] [Indexed: 12/16/2022]
Abstract
In recent years, the cancer survival of patients has improved thanks to advances in the pharmacological field. In many guidelines, cardiotoxicity induced by anticancer drugs was defined as a reduction from baseline in the left ventricular ejection fraction (LVEF) assessed by echocardiography. It is known that LVEF is not a sensible parameter in the detection of cardiotoxicity. Therefore, a decrease from baseline in the global longitudinal strain (GLS) or troponins elevation is used to detect subclinical cardiotoxicity. LVEF and GLS as well as the increase in some biomarkers are influenced by loading conditions that are frequent during chemotherapy. Other parameters not influenced by loading conditions should be used in the early diagnosis of cardiotoxicity. The aim of this review is to delineate the role of current strategies used in the early diagnosis of cardiotoxicity and to identify new strategies that could have greater application in the future in cardioncology.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Department of Health Promotion Sciences, Maternal-Infant Care, Internal Medicine and Specialties of Excellence "G. D'Alessandro ", University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy.
| | - Girolamo Manno
- Department of Health Promotion Sciences, Maternal-Infant Care, Internal Medicine and Specialties of Excellence "G. D'Alessandro ", University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - Giuseppina Novo
- Department of Health Promotion Sciences, Maternal-Infant Care, Internal Medicine and Specialties of Excellence "G. D'Alessandro ", University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| |
Collapse
|
30
|
Assessment and Management of Cardiotoxicity in Hematologic Malignancies. DISEASE MARKERS 2021; 2021:6616265. [PMID: 33613788 PMCID: PMC7875649 DOI: 10.1155/2021/6616265] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/05/2021] [Accepted: 01/27/2021] [Indexed: 12/28/2022]
Abstract
With the increasing overall survival of cancer patients due to recent discoveries in oncology, the incidence of side effects is also rising, and along with secondary malignancies, cardiotoxicity is one of the most concerning side effects, affecting the quality of life of cancer survivors. There are two types of cardiotoxicity associated with chemotherapy; the first one is acute, life-threatening but, fortunately, in most of the cases, reversible; and the second one is with late onset and mostly irreversible. The most studied drugs associated with cardiotoxicity are anthracyclines, but many new agents have demonstrated unexpected cardiotoxic effect, including those currently used in multiple myeloma treatment (proteasome inhibitors and immunomodulatory agents), tyrosine kinase inhibitors used in the treatment of chronic myeloid leukemia and some forms of acute leukemia, and immune checkpoint inhibitors recently introduced in treatment of refractory lymphoma patients. To prevent irreversible myocardial damage, early recognition of cardiac toxicity is mandatory. Traditional methods like echocardiography and magnetic resonance imaging are capable of detecting structural and functional changings, but unable to detect early myocardial damage; therefore, more sensible biomarkers like troponins and natriuretic peptides have to be introduced into the current practice. Baseline assessment of patients allows the identification of those with high risk for cardiotoxicity, while monitoring during and after treatment is important for early detection of cardiotoxicity and prompt intervention.
Collapse
|
31
|
Micheletti PL, Carla-da-Silva J, Scandolara TB, Kern R, Alves VD, Malanowski J, Victorino VJ, Herrera ACSA, Rech D, Souza JAO, Simão ANC, Panis C, Dichi I. Proinflammatory circulating markers: new players for evaluating asymptomatic acute cardiovascular toxicity in breast cancer treatment. J Chemother 2021; 33:106-115. [PMID: 33480330 DOI: 10.1080/1120009x.2021.1873632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE This study aimed to evaluate markers of cardiac damage (total CK, CKMB and CRP), inflammatory markers (free iron, homocysteine and TNF-α) as well as lipidogram in breast cancer patients undergoing acute cycles of doxorubicin (DOX), paclitaxel (PTX) or trastuzumab (TZ) and to verify if there is an association between these markers and the toxicity of the chemotherapeutic treatment. Methods: Included in the study were 120 breast cancer patients and 50 healthy controls. All analyzes were performed on automated systems. For the statistical analysis, each group was compared with the controls according to their normality by Student's t-test and Mann-Whitney test. Results: Our results showed that DOX treatment led to increased hsCRP (4.80 ± 1.23 mg/dL, p = 0.0005), triglycerides (187.6 ± 25.06, p = 0.0231), TNF-α (42.31 ± 17.96 pg/mL, p = 0.01) and Fe levels (138.8 ± 18.6 μg/dL, p = 0.0193). In the meantime, PTX induced changes in CK-MB (8.78 ± 4.2 U/L, p = 0.0361), hsCRP (7.12 ± 1.87 mg/dL, p = 0.0006), cholesterol (201.7 ± 19.54, p = 0.05), triglycerides (201.7 ± 19.54, p = 0.0277), TNF-α (38.27 ± 9.12 pg/mL, p = 0.023), homocysteine (10.95 ± 0, 86 μmol/L, p = 0.005), and free iron (113 ± 18 6 μg/dL, p = 0.045) while TZ augmented CK-MB (6.9 ± 1.97 U/L, p < 0.00), hsPCR (3.12 ± 0.68 mg/dL, p = 0.095), cholesterol (218.3 ± 16.79, p = 0.0317), triglycerides (218.3 ± 16.79, p = 0.0127), TNF-α (89.6 ± 12.11, p = 0.032), homocysteine (9.95 ± 1.15 μmol/L, p = 0.0396), free iron (120.5 ± 4.64 μg/dl, p = 0.0058) as well. Conclusions: Our data demonstrated the existence of a proinflammatory net triggered by breast cancer chemotherapy that could increase cardiomyocytes permeability and allow the leakage of circulating proteins as CK-MB and induce the production of hsCRP.
Collapse
Affiliation(s)
- P L Micheletti
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Estadual de Londrina, Paraná, Brazil
| | - J Carla-da-Silva
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - T B Scandolara
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - R Kern
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - V D Alves
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - J Malanowski
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - V J Victorino
- Instituto Federal do Rio de Janeiro, Campus Engenheiro Paulo de Frontin, Rio de Janeiro, Brazil
| | - A C S A Herrera
- Pontifícia Universidade Católica de Londrina, Paraná, Brazil
| | - D Rech
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil.,Hospital de Câncer de Francisco Beltrão, Ceonc, Paraná, Brazil
| | - J A O Souza
- Hospital de Câncer de Francisco Beltrão, Ceonc, Paraná, Brazil
| | - A N C Simão
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Estadual de Londrina, Paraná, Brazil
| | - C Panis
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - I Dichi
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Estadual de Londrina, Paraná, Brazil
| |
Collapse
|
32
|
Gonciar D, Mocan L, Zlibut A, Mocan T, Agoston-Coldea L. Cardiotoxicity in HER2-positive breast cancer patients. Heart Fail Rev 2021; 26:919-935. [PMID: 33405000 DOI: 10.1007/s10741-020-10072-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 01/22/2023]
Abstract
Due to the recent advances in diagnosis and management of patients with HER2-positive breast cancer, especially through novel HER2-targeted agents, cardiotoxicity becomes an emerging problem. Although chemotherapy significantly increases survival, the risk of cardiovascular disease development is high and still underestimated and could imply treatment discontinuation. Frequently, due to lack of rigorous diagnosis strategies, cardiotoxicity assessment is delayed, and, moreover, the efficacy of current therapy options in restoring heart function is questionable. For a comprehensive risk assessment, it is vital to characterize the clinical spectrum of HER2-targeted agents and anthracyclines, as well as their pathogenic pathways involved in cardiotoxicity. Advanced cardiovascular multimodal imaging and circulating biomarkers plays primary roles in early assessing cardiotoxicity and also in guiding specific preventive measures. Even though the knowledge in this field is rapidly expanding, there are still questions that arise regarding the optimal approach in terms of timing and methods. The aim of the current review aims to providean overview of currently available data.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Mocan
- 3rd Surgery Department, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Alexandru Zlibut
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Teodora Mocan
- Physiology Department, Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
33
|
Dent SF, Kikuchi R, Kondapalli L, Ismail-Khan R, Brezden-Masley C, Barac A, Fradley M. Optimizing Cardiovascular Health in Patients With Cancer: A Practical Review of Risk Assessment, Monitoring, and Prevention of Cancer Treatment-Related Cardiovascular Toxicity. Am Soc Clin Oncol Educ Book 2020; 40:1-15. [PMID: 32213102 DOI: 10.1200/edbk_286019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Advances in cancer screening and improved treatment approaches have led to an increase in survivorship and, consequently, recognition of an association between cancer treatments and the development of cardiovascular complications. In addition, as the population becomes proportionally older, comorbid cardiovascular risk factors are more prevalent in the population and compound the risk of developing cancer treatment-related cardiovascular toxicity. Cardio-oncology has emerged as a new subspecialty of medicine that provides a multidisciplinary approach, bringing together oncologists, cardiologists, and allied health care providers who are tasked with optimizing the cardiovascular health of patients exposed to potentially cardiotoxic cancer therapy. Using a case-based approach, practical advice on how to identify, monitor, and treat patients with cancer who are at risk for developing cancer treatment-related cardiovascular dysfunction is discussed. Cardiovascular risk factors (e.g., age, hypertension, diabetes) and cancer therapies (chemotherapy, targeted therapy, radiation) associated with cardiovascular toxicity are presented. Current cardiac monitoring strategies such as two- and three-dimensional echocardiography, cardiac MRI, and biomarkers (troponin and brain natriuretic peptide [BNP]) are discussed. Last, the current literature on pharmacologic (e.g., angiotensin-converting enzyme inhibitors, β-blockers, statins) and lifestyle (diet and exercise) strategies to mitigate cardiovascular toxicity during and following completion of cancer therapy are reviewed.
Collapse
Affiliation(s)
- Susan F Dent
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC
| | - Robin Kikuchi
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC
| | - Lavanya Kondapalli
- University of Colorado Health Cancer, University of Colorado, Aurora, CO
| | | | | | - Ana Barac
- MedStar Heart and Vascular Institute, Georgetown University, Washington, DC
| | - Michael Fradley
- Moffitt Cancer Center, University of South Florida, Tampa, FL
| |
Collapse
|
34
|
Pudil R, Mueller C, Čelutkienė J, Henriksen PA, Lenihan D, Dent S, Barac A, Stanway S, Moslehi J, Suter TM, Ky B, Štěrba M, Cardinale D, Cohen‐Solal A, Tocchetti CG, Farmakis D, Bergler‐Klein J, Anker MS, Von Haehling S, Belenkov Y, Iakobishvili Z, Maack C, Ciardiello F, Ruschitzka F, Coats AJ, Seferovic P, Lainscak M, Piepoli MF, Chioncel O, Bax J, Hulot J, Skouri H, Hägler‐Laube ES, Asteggiano R, Fernandez TL, Boer RA, Lyon AR. Role of serum biomarkers in cancer patients receiving cardiotoxic cancer therapies: a position statement from the
Cardio‐Oncology Study Group
of the
Heart Failure Association
and the
Cardio‐Oncology Council of the European Society of Cardiology. Eur J Heart Fail 2020; 22:1966-1983. [DOI: 10.1002/ejhf.2017] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/14/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Radek Pudil
- 1st Department Medicine – Cardioangiology Charles University Prague, Medical Faculty and University Hospital Hradec Kralove Prague Czech Republic
| | - Christian Mueller
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology University Hospital Basel, University of Basel Basel Switzerland
| | - Jelena Čelutkienė
- Clinic of Cardiac and Vascular Diseases Institute of Clinical Medicine, Faculty of Medicine, Vilnius University Vilnius Lithuania
- State Research Institute Centre For Innovative Medicine Vilnius Lithuania
| | | | - Dan Lenihan
- Cardio‐Oncology Center of Excellence Washington University in St Louis St Louis MO USA
| | - Susan Dent
- Duke Cancer Institute Duke University Durham NC USA
| | - Ana Barac
- MedStar Heart and Vascular Institute Georgetown University Washington DC USA
| | | | - Javid Moslehi
- Cardio‐Oncology Program, Department of Medicine Vanderbilt University Medical Center Nashville TN USA
| | - Thomas M. Suter
- Department of Cardiology Bern University Hospital, Inselspital, University of Bern Bern Switzerland
| | - Bonnie Ky
- University of Pennsylvania Philadelphia PA USA
| | - Martin Štěrba
- Department of Pharmacology Faculty of Medicine in Hradec Kralove, Charles University Hradec Kralove Czech Republic
| | - Daniela Cardinale
- Cardioncology Unit European Institute of Oncology, IRCCS Milan Italy
| | - Alain Cohen‐Solal
- UMR‐S 942, Paris University, Cardiology Department, Lariboisiere Hospital, AP‐HP Paris France
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center for Clinical and Translational Research (CIRCET) ‘Federico II’ University Naples Italy
| | - Dimitrios Farmakis
- University of Cyprus Medical School Nicosia Cyprus
- Cardio‐Oncology Clinic, Heart Failure Unit, ‘Attikon’ University Hospital Athens Greece
- National and Kapodistrian University of Athens Medical School Athens Greece
| | | | - Markus S. Anker
- Division of Cardiology and Metabolism, Department of Cardiology Charité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research), partner site Berlin and Department of Cardiology, Charité Campus Benjamin Franklin Berlin Germany
| | - Stephan Von Haehling
- Department of Cardiology and Pneumology University of Goettingen Medical Center Goettingen Germany
- German Center for Cardiovascular Research (DZHK), partner site Goettingen Goettingen Germany
| | | | - Zaza Iakobishvili
- Department of Community Cardiology Tel Aviv Jaffa District, Clalit Health Fund and Sackler Faculty of Medicine, Tel Aviv University Tel Aviv Israel
| | - Christoph Maack
- Comprehensive Heart Failure Center University Clinic Würzburg Würzburg Germany
| | - Fortunato Ciardiello
- Department of Precision Medicine ‘Luigi Vanvitelli’ University of Campania Naples Italy
| | - Frank Ruschitzka
- University Heart Center, Department of Cardiology University Hospital Zurich Zurich Switzerland
| | - Andrew J.S. Coats
- University of Warwick Warwick UK
- Pharmacology Centre of Clinical and Experimental Medicine, IRCCS San Raffaele Pisana Rome Italy
| | - Petar Seferovic
- Faculty of Medicine and Serbian Academy of Sciences and Arts University of Belgrade Belgrade Serbia
| | | | - Massimo F. Piepoli
- Cardiac Department ‘Guglielmo da Saliceto’ Polichirurgico Hospital AUSL Piacenza Piacenza Italy
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases ‘Prof. C.C. Iliescu’ Bucharest Romania
- University of Medicine Carol Davila Bucharest Romania
| | - Jereon Bax
- Department of Cardiology Leiden University Medical Centre Leiden The Netherlands
| | - Jean‐Sebastien Hulot
- Université de Paris CIC1418, Paris Cardiovascular Research Center, INSERM Paris France
| | - Hadi Skouri
- Cardiology Division, Internal Medicine Department at American University of Beirut Medical Center Beirut Lebanon
| | | | | | - Teresa Lopez Fernandez
- Cardiology Service Cardio‐Oncology Unit, La Paz University Hospital and IdiPAz Research Institute, Ciber CV Madrid Spain
| | - Rudolf A. Boer
- Department of Cardiology University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | - Alexander R. Lyon
- Cardio‐Oncology Service Royal Brompton Hospital and Imperial College London London UK
| |
Collapse
|
35
|
Berliner D, Beutel G, Bauersachs J. Echocardiography and biomarkers for the diagnosis of cardiotoxicity. Herz 2020; 45:637-644. [DOI: 10.1007/s00059-020-04957-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A, Herrmann J, Porter C, Lyon AR, Lancellotti P, Patel A, DeCara J, Mitchell J, Harrison E, Moslehi J, Witteles R, Calabro MG, Orecchia R, de Azambuja E, Zamorano JL, Krone R, Iakobishvili Z, Carver J, Armenian S, Ky B, Cardinale D, Cipolla CM, Dent S, Jordan K. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol 2020; 31:171-190. [PMID: 31959335 PMCID: PMC8019325 DOI: 10.1016/j.annonc.2019.10.023] [Citation(s) in RCA: 543] [Impact Index Per Article: 135.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer and cardiovascular (CV) disease are the most prevalent diseases in the developed world. Evidence increasingly shows that these conditions are interlinked through common risk factors, coincident in an ageing population, and are connected biologically through some deleterious effects of anticancer treatment on CV health. Anticancer therapies can cause a wide spectrum of short- and long-term cardiotoxic effects. An explosion of novel cancer therapies has revolutionised this field and dramatically altered cancer prognosis. Nevertheless, these new therapies have introduced unexpected CV complications beyond heart failure. Common CV toxicities related to cancer therapy are defined, along with suggested strategies for prevention, detection and treatment. This ESMO consensus article proposes to define CV toxicities related to cancer or its therapies and provide guidance regarding prevention, screening, monitoring and treatment of CV toxicity. The majority of anticancer therapies are associated with some CV toxicity, ranging from asymptomatic and transient to more clinically significant and long-lasting cardiac events. It is critical however, that concerns about potential CV damage resulting from anticancer therapies should be weighed against the potential benefits of cancer therapy, including benefits in overall survival. CV disease in patients with cancer is complex and treatment needs to be individualised. The scope of cardio-oncology is wide and includes prevention, detection, monitoring and treatment of CV toxicity related to cancer therapy, and also ensuring the safe development of future novel cancer treatments that minimise the impact on CV health. It is anticipated that the management strategies discussed herein will be suitable for the majority of patients. Nonetheless, the clinical judgment of physicians remains extremely important; hence, when using these best clinical practices to inform treatment options and decisions, practitioners should also consider the individual circumstances of their patients on a case-by-case basis.
Collapse
Affiliation(s)
- G. Curigliano
- European Institute of Oncology IRCCS, Milan
- Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - D. Lenihan
- Cardiovascular Division, Cardio-Oncology Center of Excellence, Washington University Medical Center, St. Louis
| | - M. Fradley
- Cardio-oncology Program, Division of Cardiovascular Medicine, Morsani College of Medicine and H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa
| | - S. Ganatra
- Cardio-Oncology Program, Lahey Medical Center, Burlington
| | - A. Barac
- Cardio-Oncology Program, Medstar Heart and Vascular Institute and MedStar Georgetown Cancer Institute, Georgetown University Hospital, Washington DC
| | - A. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis
| | | | - C. Porter
- University of Kansas Medical Center, Lawrence, USA
| | - A. R. Lyon
- Royal Brompton Hospital and Imperial College, London, UK
| | - P. Lancellotti
- GIGA Cardiovascular Sciences, Acute Care Unit, Heart Failure Clinic, CHU Sart Tilman, University Hospital of Liège, Liège, Belgium
| | - A. Patel
- Morsani College of Medicine, University of South Florida, Tampa
| | - J. DeCara
- Medicine Section of Cardiology, University of Chicago, Chicago
| | - J. Mitchell
- Washington University Medical Center, St. Louis
| | - E. Harrison
- HCA Memorial Hospital and University of South Florida, Tampa
| | - J. Moslehi
- Vanderbilt University School of Medicine, Nashville
| | - R. Witteles
- Division of Cardiovascular Medicine, Falk CVRC, Stanford University School of Medicine, Stanford, USA
| | - M. G. Calabro
- Department of Anesthesia and Intensive Care, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | | | - E. de Azambuja
- Institut Jules Bordet and L’Université Libre de Bruxelles, Brussels, Belgium
| | | | - R. Krone
- Division of Cardiology, Washington University, St. Louis, USA
| | - Z. Iakobishvili
- Clalit Health Services, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J. Carver
- Division of Cardiology, Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia
| | - S. Armenian
- Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte
| | - B. Ky
- University of Pennsylvania School of Medicine, Philadelphia, USA
| | - D. Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan
| | - C. M. Cipolla
- Cardiology Department, European Institute of Oncology, IRCCS, Milan, Italy
| | - S. Dent
- Duke Cancer Institute, Duke University, Durham, USA
| | - K. Jordan
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - ESMO Guidelines Committee
- Correspondence to: ESMO Guidelines Committee, ESMO Head Office, Via Ginevra 4, CH-6900 Lugano, Switzerland, (ESMO Guidelines Committee)
| |
Collapse
|
37
|
Demissei BG, Hubbard RA, Zhang L, Smith AM, Sheline K, McDonald C, Narayan V, Domchek SM, DeMichele A, Shah P, Clark AS, Fox K, Matro J, Bradbury AR, Knollman H, Getz KD, Armenian SH, Januzzi JL, Tang WHW, Liu P, Ky B. Changes in Cardiovascular Biomarkers With Breast Cancer Therapy and Associations With Cardiac Dysfunction. J Am Heart Assoc 2020; 9:e014708. [PMID: 31959034 PMCID: PMC7033834 DOI: 10.1161/jaha.119.014708] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background We examined the longitudinal associations between changes in cardiovascular biomarkers and cancer therapy–related cardiac dysfunction (CTRCD) in patients with breast cancer treated with cardotoxic cancer therapy. Methods and Results Repeated measures of high‐sensitivity cardiac troponin T (hs‐cTnT), NT‐proBNP (N‐terminal pro‐B‐type natriuretic peptide), myeloperoxidase, placental growth factor, and growth differentiation factor 15 were assessed longitudinally in a prospective cohort of 323 patients treated with anthracyclines and/or trastuzumab followed over a maximum of 3.7 years with serial echocardiograms. CTRCD was defined as a ≥10% decline in left ventricular ejection fraction to a value <50%. Associations between changes in biomarkers and left ventricular ejection fraction were evaluated in repeated‐measures linear regression models. Cox regression models assessed the associations between biomarkers and CTRCD. Early increases in all biomarkers occurred with anthracycline‐based regimens. hs‐cTnT levels >14 ng/L at anthracycline completion were associated with a 2‐fold increased CTRCD risk (hazard ratio, 2.01; 95% CI, 1.00–4.06). There was a modest association between changes in NT‐proBNP and left ventricular ejection fraction in the overall cohort; this was most pronounced with sequential anthracycline and trastuzumab (1.1% left ventricular ejection fraction decline [95% CI, −1.8 to –0.4] with each NT‐proBNP doubling). Increases in NT‐proBNP were also associated with CTRCD (hazard ratio per doubling, 1.56; 95% CI, 1.32–1.84). Increases in myeloperoxidase were associated with CTRCD in patients who received sequential anthracycline and trastuzumab (hazard ratio per doubling, 1.28; 95% CI, 1.04–1.58). Conclusions Cardiovascular biomarkers may play an important role in CTRCD risk prediction in patients with breast cancer who receive cardiotoxic cancer therapy, particularly in those treated with sequential anthracycline and trastuzumab therapy. Clinical Trial Registration URL: https://www.clinicaltrials.gov/. Unique identifier: NCT01173341.
Collapse
Affiliation(s)
- Biniyam G Demissei
- Department of Medicine Division of Cardiology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Rebecca A Hubbard
- Department of Biostatistics, Epidemiology & Informatics Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Liyong Zhang
- Division of Cardiology University of Ottawa Heart Institute Ottawa Ontario Canada
| | - Amanda M Smith
- Department of Medicine Division of Cardiology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Karyn Sheline
- Department of Medicine Division of Cardiology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Caitlin McDonald
- Department of Medicine Division of Cardiology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Vivek Narayan
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Susan M Domchek
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Angela DeMichele
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Payal Shah
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Amy S Clark
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Kevin Fox
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Jennifer Matro
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Angela R Bradbury
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Hayley Knollman
- Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Medicine Division of Hematology and Oncology University of Pennsylvania Philadelphia PA
| | - Kelly D Getz
- Division of Oncology The Children's Hospital of Philadelphia PA
| | | | - James L Januzzi
- Division of Cardiovascular Medicine Massachusetts General Hospital Boston MA
| | - W H Wilson Tang
- Division of Cardiovascular Medicine Cleveland Clinic Cleveland OH
| | - Peter Liu
- Division of Cardiology University of Ottawa Heart Institute Ottawa Ontario Canada
| | - Bonnie Ky
- Department of Medicine Division of Cardiology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Department of Biostatistics, Epidemiology & Informatics Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| |
Collapse
|
38
|
Abstract
Cardiotoxicity is a known complication of many cancer therapies. While the cardiotoxicity of established agents such as anthracyclines, antimetabolites, and alkylating agents is well known, it is important to realize that newer anticancer therapies such as tyrosine kinase inhibitors, angiogenesis inhibitors, and checkpoint inhibitors are also associated with significant adverse cardiovascular effects. Echocardiography, magnetic resonance imaging, and radionuclide imaging have been used to identify these complications early and prevent further consequences. We will discuss the different classes of cancer therapeutic agents that cause cardiotoxicity, the mechanisms that lead to these effects, and strategies that can be used to prevent the cardiac morbidity and mortality associated with their use.
Collapse
|
39
|
Lenihan DJ, Fradley MG, Dent S, Brezden-Masley C, Carver J, Filho RK, Neilan TG, Blaes A, Melloni C, Herrmann J, Armenian S, Thavendiranathan P, Armstrong GT, Ky B, Hajjar L. Proceedings From the Global Cardio-Oncology Summit: The Top 10 Priorities to Actualize for CardioOncology. JACC CardioOncol 2019; 1:256-272. [PMID: 34396188 PMCID: PMC8352295 DOI: 10.1016/j.jaccao.2019.11.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/04/2019] [Indexed: 12/27/2022] Open
Abstract
The discipline of cardio-oncology has expanded at a remarkable pace. Recent developments and challenges to clinicians who practice cardio-oncology were presented at the Global Cardio-Oncology Summit on October 3 to 4, 2019, in São Paulo, Brazil. Here, we present the top 10 priorities for our field that were discussed at the meeting, and also detail a potential path forward to address these challenges. Defining robust predictors of cardiotoxicity, clarifying the role of cardioprotection, managing and preventing thromboembolism, improving hematopoietic stem cell transplant outcomes, personalizing cardiac interventions, building the cardio-oncology community, detecting and treating cardiovascular events associated with immunotherapy, understanding tyrosine kinase inhibitor cardiotoxicity, and enhancing survivorship care are all priorities for the field. The path forward requires a commitment to research, education, and excellence in clinical care to improve our patients' lives.
Collapse
Key Words
- CV, cardiovascular
- CVD, cardiovascular disease
- DOAC, direct oral anticoagulant
- GCOS, Global Cardio-Oncology Summit
- GLS, global longitudinal strain
- HCT, hematopoietic cell transplantation
- ICI, immune checkpoint inhibitor
- LVEF, left ventricular ejection fraction
- PD-1, programmed cell death 1 or its ligand
- PD-L1, programmed cell death ligand 1
- TKI, tyrosine kinase inhibitor
- VTE, venous thromboembolism
- anthracycline
- antiangiogenic therapy
- bone marrow transplantation
- breast cancer
- cancer survivorship
- immunotherapy
- thrombosis
- tyrosine kinase inhibitor
Collapse
Affiliation(s)
- Daniel J. Lenihan
- Cardio-Oncology Center of Excellence, Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Michael G. Fradley
- Cardio-Oncology Program, Moffitt Cancer Center, University of South Florida, Tampa, Florida, USA
| | - Susan Dent
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Joseph Carver
- Cardio-Oncology Center of Excellence at the Abramson Cancer Center, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roberto Kalil Filho
- Cardio-Oncology Program, Department of Cardiopneumology, Cancer Institute and Heart Institute, University of São Paulo, Brazil
| | - Tomas G. Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne Blaes
- Division of Hematology/Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chiara Melloni
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Saro Armenian
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, University Health Network, University of Toronto, Toronto, Canada
| | - Gregory T. Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Bonnie Ky
- Cardio-Oncology Center of Excellence at the Abramson Cancer Center, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ludhmila Hajjar
- Cardio-Oncology Program, Department of Cardiopneumology, Cancer Institute and Heart Institute, University of São Paulo, Brazil
| |
Collapse
|
40
|
Lu X, Zhao Y, Chen C, Han C, Xue L, Xing D, Huang O, Tao M. BNP as a marker for early prediction of anthracycline-induced cardiotoxicity in patients with breast cancer. Oncol Lett 2019; 18:4992-5001. [PMID: 31612011 DOI: 10.3892/ol.2019.10827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Anthracycline chemotherapy serves an important role in treating patients with breast cancer but is associated with cardiotoxicity. Although brain natriuretic peptide (BNP) is not the ideal marker for detecting the presence of diseases of the heart, several studies have demonstrated the predictive utility of BNP in the diagnosis of anthracycline-induced cardiotoxicity (AIC). The aim of the present study was to evaluate the role of BNP as a marker for the early prediction of AIC in patients with breast cancer. In the present study, a total of 149 patients with breast cancer who received anthracycline therapy were evaluated. The levels of BNP and echocardiography were detected during the anthracycline-based chemotherapy and patients were followed up after chemotherapy to determine the cardiotoxicity-free survival times. In the patients who received chemotherapy, an increase in the levels of BNP was observed. The concentration of BNP was significantly higher in the cardiotoxicity group during anthracycline chemotherapy (P=0.022) compared with the non-cardiotoxicity group and it was an independent predictor of cardiotoxicity (P=0.028). The optimal diagnostic threshold of BNP after the last anthracycline chemotherapy treatment was 107.9 pg/ml, the diagnostic sensitivity was 0.538, the specificity was 0.794, the Youden index was 0.332, the positive predictive value was 0.583 and the negative predictive value was 0.762. Based on the BNP threshold, a log-rank test was performed and it was determined that the cardiotoxicity-free survival rate of the group with higher levels of BNP was always lower compared with the group with lower levels of BNP. BNP elevation was associated with cardiotoxicity during the anthracycline chemotherapy. Detecting BNP after the final treatment of anthracycline chemotherapy may contribute to the early detection of cardiotoxicity.
Collapse
Affiliation(s)
- Xiang Lu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Department of Breast Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314001, P.R. China
| | - Yingying Zhao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Caiping Chen
- Department of Breast Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314001, P.R. China
| | - Chao Han
- Department of Breast Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314001, P.R. China
| | - Li Xue
- Department of Breast Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314001, P.R. China
| | - Dan Xing
- Department of Breast Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314001, P.R. China
| | - Ou Huang
- Comprehensive Breast Health Centre, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
41
|
Hui D, Naberhuis J, Dibaj S, Naqvi M, Liu D, Bruera E. Association Between Plasma Brain Natriuretic Peptide and Overall Survival in Patients With Advanced Cancer: Preliminary Findings. J Pain Symptom Manage 2019; 58:465-471. [PMID: 31128228 PMCID: PMC6708736 DOI: 10.1016/j.jpainsymman.2019.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/12/2019] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
CONTEXT Atrial and brain natriuretic peptides (ANP and BNP) are established diagnostic and prognostic markers in heart failure, but their utility in patients with advanced cancer is unclear. OBJECTIVES Our objective was to examine the association between plasma natriuretic peptides and survival in patients with advanced cancer without clinical evidence of heart failure. METHODS This exploratory analysis of a multicenter, randomized clinical trial of cancer patients receiving hospice care assessed the association between elevated plasma ANP, BNP, or Pro-BNP (cutoffs of >77, 100, and 900 pg/mL, respectively) and overall survival. Time-to-event analyses, including multivariate Cox regression, were conducted. RESULTS Among 97 patients, the mean age was 67.2 years and the overall survival was 16 days (95% CI, 13-23 days). ANP, BNP, and Pro-BNP were elevated in 29 of 36 (81%), nine of 23 (39%), and 32 of 38 (84%) patients, respectively. Elevated ANP, BNP, or Pro-BNP was associated with worse survival (median 14 vs. 21 days; P = 0.02). BNP or Pro-BNP was inversely associated with overall survival (hazard ratio = 2.27; 95% CI, 1.29-3.97) in univariate Cox regression analysis, and remained significant in multivariate Cox regression analysis (hazard ratio = 3.09; 95% CI, 1.40-6.84) after adjusting for treatment group and known prognostic variables such as performance status, albumin, creatinine, delirium, dyspnea, and anorexia. Elevated ANP alone was not significantly associated with survival (P = 0.17). CONCLUSION Our preliminary findings suggest that BNP or Pro-BNP may be a novel objective prognostic marker in cancer patients without heart failure. Further research is needed to confirm these findings.
Collapse
Affiliation(s)
- David Hui
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Jane Naberhuis
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Seyedeh Dibaj
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mujtaba Naqvi
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Diane Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eduardo Bruera
- Department of Palliative Care, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
42
|
Di JY, Zhang ZX, Xin SJ. Glycogen Phosphorylase Isoenzyme Bb, Myoglobin and BNP in ANT-Induced Cardiotoxicity. Open Life Sci 2018; 13:561-568. [PMID: 33817127 PMCID: PMC7874740 DOI: 10.1515/biol-2018-0067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022] Open
Abstract
Anthracyline (ANT) has been demonstrated as a useful treatment for leukemia and solid tumors. However, ANT has previously reported cardiotoxic effects, which can reduce the therapeutic index for cancer treatment. This study aimed to investigate the associations of glycogen phosphorylase isoenzyme BB (GPBB), myoglobin (Mb), and brain natriuretic peptide (BNP) with anthracycline (ANT-induced cardiotoxicity (AIC)) amongst the Chinese population. Patients suffering from leukemia were recruited. Electrocardiogram and echocardiography were used along with chemotherapy to determine left ventricular ejection fraction (LVEF), mitral ratio of peak early to late diastolic filling velocity (E/A), E-wave deceleration time (EDT), and isovolumic relaxation time (IVRT). Double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA) was employed to examine and compare serum GPBB, Mb, and BNP levels. Following chemotherapy, the patients presented higher levels of serum GPBB, Mb, and BNP than before chemotherapy treatment. The levels of LVEF (%), E/A, and IVRT were significantly decreased after chemotherapy, while EDT was markedly increased. The cumulative ANT dose was positively corelated to serum GPBB, Mb, and BNP levels while it was negatively corelated to LVEF levels. In conclusion, serum GPBB, Mb, and BNP levels in combination might provide higher diagnostic accuracy in the early detection of AIC compared with other single indicators.
Collapse
Affiliation(s)
- Jia-Yin Di
- Clinical Laboratory, Department of Outpatient, Huzhou University, Huzhou 313000, Huzhou, P.R. China
| | - Zong-Xin Zhang
- Department of Clinical Laboratory, Huzhou Central Hospital, No. 198, Hongqi Road, Huzhou 313000, Zhejiang Province, P.R. China
| | - Shao-Jun Xin
- Department of Clinical Laboratory, Huzhou Central Hospital, No. 198, Hongqi Road, Huzhou 313000, Zhejiang Province, P.R. China
| |
Collapse
|
43
|
Nakanishi K, Daimon M. Early Prediction of Cardiotoxicity From Cancer Chemotherapy ― Can Baseline Strain Identify High-Risk Patients? ―. Circ J 2018; 82:2477-2478. [DOI: 10.1253/circj.cj-18-0836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Koki Nakanishi
- Department of Cardiovascular Medicine, The University of Tokyo
| | - Masao Daimon
- Department of Cardiovascular Medicine, The University of Tokyo
- Department of Clinical Laboratory, The University of Tokyo
| |
Collapse
|
44
|
The role of cardiac biomarkers in cardio-oncology. Curr Probl Cancer 2018; 42:375-385. [PMID: 30126650 DOI: 10.1016/j.currproblcancer.2018.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 11/24/2022]
Abstract
Major advances in cancer therapy have resulted in an unprecedented improvement in patient survival for many cancers. Partially as a consequence of increased longevity, it has become evident that the cardiovascular system is frequently impacted by chemotherapy, radiation, and particularly certain targeted therapy. Cardiotoxicity continues to be a dose-limiting side effect of many chemotherapeutic agents including the anthracyclines. Early identification of patients at increased risk of cardiotoxicity or detecting cardiac injury at the earliest point will allow for initiation of cardio-protective strategies and hopefully prevent or reduce the need to modify life-saving cancer therapies. Biomarkers offer a potential solution to this clinical challenge because they may assist us in early identification of subclinical cardiac disease. In this review, we will summarize studies conducted evaluating the use of cardiac troponins (TnI, TnT) and natriuretic peptides, BNP and NT-proBNP. Other biomarkers for cardiotoxicity that have been at least preliminarily investigated, including myeloperoxidase and microRNAs, will be discussed as an exploratory tool for evidence of cardiac dysfunction during cancer treatment.
Collapse
|
45
|
|
46
|
Abstract
Biomarkers are at the cornerstone of preventive measures and contribute to the screening process. More recently, biomarkers have been used to gauge the biological response to the employed therapies. Since it is ubiquitously used to detect subclinical disease process, biomarkers also have found its place in cancer therapy related cardiac dysfunction (CTRCD). The aim of this review is to comprehensively present up-to-date knowledge of biomarkers in CTRCD and highlight some of the future biomedical technologies that may strengthen the screening process, and/or provide new insight in pathological mechanisms behind CTRCD.
Collapse
Affiliation(s)
- Rohit Moudgil
- Division of Cardiology, MD Anderson Cancer Center, 1515 W Holcombe Blvd, Houston, TX, 77030, USA.
| | - Parag A Parekh
- Department of Endocrine Neoplasia and Hormonal Disorders, MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
47
|
Kirkham A, Shave R, Bland K, Bovard J, Eves N, Gelmon K, McKenzie D, Virani S, Stöhr E, Warburton D, Campbell K. Protective effects of acute exercise prior to doxorubicin on cardiac function of breast cancer patients: A proof-of-concept RCT. Int J Cardiol 2017; 245:263-270. [DOI: 10.1016/j.ijcard.2017.07.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/23/2017] [Accepted: 07/12/2017] [Indexed: 01/16/2023]
|
48
|
Shim JV, Chun B, van Hasselt JGC, Birtwistle MR, Saucerman JJ, Sobie EA. Mechanistic Systems Modeling to Improve Understanding and Prediction of Cardiotoxicity Caused by Targeted Cancer Therapeutics. Front Physiol 2017; 8:651. [PMID: 28951721 PMCID: PMC5599787 DOI: 10.3389/fphys.2017.00651] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are highly potent cancer therapeutics that have been linked with serious cardiotoxicity, including left ventricular dysfunction, heart failure, and QT prolongation. TKI-induced cardiotoxicity is thought to result from interference with tyrosine kinase activity in cardiomyocytes, where these signaling pathways help to control critical processes such as survival signaling, energy homeostasis, and excitation–contraction coupling. However, mechanistic understanding is limited at present due to the complexities of tyrosine kinase signaling, and the wide range of targets inhibited by TKIs. Here, we review the use of TKIs in cancer and the cardiotoxicities that have been reported, discuss potential mechanisms underlying cardiotoxicity, and describe recent progress in achieving a more systematic understanding of cardiotoxicity via the use of mechanistic models. In particular, we argue that future advances are likely to be enabled by studies that combine large-scale experimental measurements with Quantitative Systems Pharmacology (QSP) models describing biological mechanisms and dynamics. As such approaches have proven extremely valuable for understanding and predicting other drug toxicities, it is likely that QSP modeling can be successfully applied to cardiotoxicity induced by TKIs. We conclude by discussing a potential strategy for integrating genome-wide expression measurements with models, illustrate initial advances in applying this approach to cardiotoxicity, and describe challenges that must be overcome to truly develop a mechanistic and systematic understanding of cardiotoxicity caused by TKIs.
Collapse
Affiliation(s)
- Jaehee V Shim
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| | - Bryan Chun
- Department of Biomedical Engineering, University of VirginiaCharlottesville, VA, United States
| | - Johan G C van Hasselt
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| | - Marc R Birtwistle
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of VirginiaCharlottesville, VA, United States
| | - Eric A Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, United States
| |
Collapse
|
49
|
Cardiac Imaging: Multimodality Advances and Surveillance Strategies in Detection of Cardiotoxicity. Curr Oncol Rep 2017; 19:63. [PMID: 28791609 DOI: 10.1007/s11912-017-0622-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Contemporary cancer management has increased the overall number of cancer survivors, but cardiotoxicity remains a subject of concern, which is a major cause of noncancer mortality among survivors. Among the potential cardiovascular complications, left ventricular (LV) systolic dysfunction is a poor prognostic factor. The importance of its early detection is based on the principle that the likelihood of response to heart failure (HF) treatment is temporally related to the initiation of HF treatment. For these reasons, cardiac monitoring is commonly applied in general practice, based on serial measurements of LV ejection fraction (LVEF); transthoracic echocardiography (TTE) is generally used. However, the LVEF, as a diagnostic and predictive parameter, has significant limitations, which calls for more effective multimodality imaging strategies. This approach requires further study, but there is increasing available data in the literature, encouraging the combination of multimodality imaging parameters and techniques for early cancer therapeutic-related cardiac dysfunction (CTRCD) detection.
Collapse
|
50
|
Levis BE, Binkley PF, Shapiro CL. Cardiotoxic effects of anthracycline-based therapy: what is the evidence and what are the potential harms? Lancet Oncol 2017; 18:e445-e456. [DOI: 10.1016/s1470-2045(17)30535-1] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/04/2017] [Accepted: 01/12/2017] [Indexed: 01/07/2023]
|